1
|
Sethy B, Upadhyay R, Narwanti I, Yu ZY, Lee SB, Liou JP. Novel dual inhibitor targeting CDC25 and HDAC for treating triple-negative breast cancer. Apoptosis 2024; 29:2047-2073. [PMID: 39395083 PMCID: PMC11550225 DOI: 10.1007/s10495-024-02023-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/14/2024]
Abstract
Triple-negative breast cancer (TNBC) presents a significant challenge for treatment due to its aggressive nature and the lack of effective therapies. This study developed dual inhibitors against cell division cycle 25 (CDC25) and histone deacetylases (HDACs) for TNBC treatment. CDC25 phosphatases are crucial for activating cyclin-dependent kinases (CDKs), the master regulators of cell cycle progression. HDACs regulate various biological processes by deacetylating histone and non-histone proteins, affecting gene expression, chromatin structure, cell differentiation, and proliferation. Dysregulations of HDAC and CDC25 are associated with several human malignancies. We generated a group of dual inhibitors for CDC25 and HDAC by combining the molecular structures of CDC25 (quinoline-5,8-dione) and HDAC (hydroxamic acid or benzamide) pharmacophores. The newly developed compounds were evaluated against various solid-tumor, leukemia, and non-malignant breast epithelial cells. Among the synthesized compounds, 18A emerged as a potent inhibitor, demonstrating significant cytotoxicity against TNBC cells, superior to its effects on other cancer types while sparing non-malignant cells. 18A possessed similar HDAC inhibitory activity as MS-275 and potently suppressed CDC25 activity in vitro and the CDK1 dephosphorylation in cells. Additionally, 18A hindered the progression of S and G2/M phases, triggered DNA damage, and induced apoptosis. These findings underscore the potential of 18A as a targeted therapy for TNBC and warrants further preclinical development.
Collapse
Affiliation(s)
- Bidyadhar Sethy
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Richa Upadhyay
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Iin Narwanti
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Faculty of Pharmacy, Universitas Ahmad Dahlan, Yogyakarta, Indonesia
| | - Zih-Yao Yu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sung-Bau Lee
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Abrams JN. Design, Synthesis, and In Vitro Mitotic Evaluation of 3‐Amino‐Isoquinolinones as Anticancer Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202202861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jason N. Abrams
- Department of Chemistry Texas A&M University Kingsville 700 University Blvd Kingsville TX 78363
| |
Collapse
|
3
|
Hernandez CC, Tarfa RA, Miguel I Limcaoco J, Liu R, Mondal P, Hill C, Keith Duncan R, Tzounopoulos T, Stephenson CRJ, O'Meara MJ, Wipf P. Development of an automated screen for Kv7.2 potassium channels and discovery of a new agonist chemotype. Bioorg Med Chem Lett 2022; 71:128841. [PMID: 35671848 PMCID: PMC9469649 DOI: 10.1016/j.bmcl.2022.128841] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 01/11/2023]
Abstract
To identify pore domain ligands on Kv7.2 potassium ion channels, we compared wild-type (WT) and W236L mutant Kv7.2 channels in a series of assays with previously validated and novel agonist chemotypes. Positive controls were retigabine, flupirtine, and RL-81; i.e. Kv7.2 channel activators that significantly shift voltage-dependent activation to more negative potentials (ΔV50) at 5 µM. We identified 6 new compounds that exhibited differential enhancing activity between WT and W236L mutant channels. Whole cell patch-clamp electrophysiology studies were conducted to identify Kv7.2. Kv7.2/3, Kv7.4, and Kv7.5 selectivity. Our results validate the SyncroPatch platform and establish new structure activity relationships (SAR). Specifically, in addition to selective Kv7.2, Kv7.2/3, Kv7.4. and Kv7.5 agonists, we identified a novel chemotype, ZK-21, a 4-aminotetrahydroquinoline that is distinct from any of the previously described Kv7 channel modifiers. Using flexible receptor docking, ZK-21 was predicted to be stabilized by W236 and bind perpendicular to retigabine, burying the benzyl carbamate group into a tunnel reaching the core of the pore domain.
Collapse
Affiliation(s)
- Ciria C Hernandez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Rahilla A Tarfa
- Department of Otolaryngology, Pittsburgh Hearing Research Center, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jose Miguel I Limcaoco
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ruiting Liu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Pravat Mondal
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Clare Hill
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - R Keith Duncan
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Thanos Tzounopoulos
- Department of Otolaryngology, Pittsburgh Hearing Research Center, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Corey R J Stephenson
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, United States; School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
4
|
Yu J, Yao HW, Liu TT, Wang D, Shi JH, Yuan GW, Ma S, Wu LY. Comprehensive Analysis and Experimental Validation of a Novel Estrogen/Progesterone-Related Prognostic Signature for Endometrial Cancer. J Pers Med 2022; 12:jpm12060914. [PMID: 35743699 PMCID: PMC9225250 DOI: 10.3390/jpm12060914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Estrogen and progesterone are the major determinants of the occurrence and development of endometrial cancer (EC), which is one of the most common gynecological cancers worldwide. Our purpose was to develop a novel estrogen/progesterone-related gene signature to better predict the prognosis of EC and help discover effective therapeutic strategies. We downloaded the clinical and RNA-seq data of 397 EC patients from The Cancer Genome Atlas (TCGA) database. The “limma” R package was used to screen for estrogen/progesterone-related differentially expressed genes (DEGs) between EC and normal tissues. Univariate and multivariate Cox proportional hazards regression analyses were applied to identify these DEGs that were associated with prognosis; then, a novel estrogen/progesterone-related prognostic signature comprising CDC25B, GNG3, ITIH3, PRXL2A and SDHB was established. The Kaplan–Meier (KM) survival analysis showed that the low-risk group identified by this signature had significantly longer overall survival (OS) than the high-risk group; the receiver operating characteristic (ROC) and risk distribution curves suggested this signature was an accurate predictor independent of risk factors. A nomogram incorporating the signature risk score and stage was constructed, and the calibration plot suggested it could accurately predict the survival rate. Compared with normal tissues, tumor tissues had increased mRNA levels of GNG3 and PRXL2A and a reduced mRNA level of ITIH3. The knockdown of PRXL2A and GNG3 significantly inhibited the proliferation and colony formation of Ishikawa and AN3CA cells, while the inhibition of PRXL2A expression suppressed xenograft growth. In this study, five estrogen/progesterone-related genes were identified and incorporated into a novel signature, which provided a new classification tool for improved risk assessment and potential molecular targets for EC therapies.
Collapse
Affiliation(s)
- Jing Yu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Hong-Wen Yao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Ting-Ting Liu
- Department of Blood Grouping, Beijing Red Cross Blood Center, Beijing 100088, China;
| | - Di Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China;
| | - Jian-Hong Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China;
| | - Guang-Wen Yuan
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Sai Ma
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
- Correspondence: (S.M.); (L.-Y.W.)
| | - Ling-Ying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
- Correspondence: (S.M.); (L.-Y.W.)
| |
Collapse
|
5
|
Ribeiro HF, de Castro Sant' Anna C, de Jesus Oliveira Kato V, de Sousa Brasil RM, Bona AB, da Costa DF, Lima IK, Soares PC, Guimarães APA, de Assumpção PP, Burbano RR. CDC25B Inhibition by menadione: a potential new therapeutical approach. Anticancer Agents Med Chem 2022; 22:2927-2932. [PMID: 35440317 DOI: 10.2174/1871520622666220418131935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/28/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is the fifth most common type of tumor and the third leading cause of cancer death worldwide. The evolution of gastric carcinogenesis is still poorly understood and, for this reason, preclinical research protocols were established that included the development of gastric cancer cell lines and the establishment of models of gastric carcinogenesis in non-human primate Sapajus apella. A comprehensive literature search was performed in relevant databases such as PubMed, ResearchGate and Google Scholar to identify studies related to the topic. After an in-depth study of these reports, significant data/data were collected and compiled under appropriate headings. The main result of the studies carried out by the group on GC is the demonstration of the MYC gene overexpression as a common phenomenon in stomach carcinogenesis. Furthermore, we revealed that reducing the expression of the CDC25B gene, regulated by the MYC protein, is a therapeutic strategy against stomach tumors. This review article reveals preclinical evidence that treatment with menadione in experimental models of gastric tumorigenesis, in vivo and in vitro, inhibits the action of the phosphatase CDC25B and, consequently, prevents cell proliferation, invasion and migration.
Collapse
|
6
|
Sangwan S, Yadav N, Kumar R, Chauhan S, Dhanda V, Walia P, Duhan A. A score years’ update in the synthesis and biological evaluation of medicinally important 2-pyridones. Eur J Med Chem 2022; 232:114199. [DOI: 10.1016/j.ejmech.2022.114199] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/10/2022] [Indexed: 12/18/2022]
|
7
|
Kim IH, Eom T, Park JY, Kim HJ, Nam TJ. Dichloromethane fractions of Calystegia soldanella induce S‑phase arrest and apoptosis in HT‑29 human colorectal cancer cells. Mol Med Rep 2021; 25:60. [PMID: 34935054 PMCID: PMC8767546 DOI: 10.3892/mmr.2021.12576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022] Open
Abstract
Calystegia soldanella is a halophyte and a perennial herb that grows on coastal sand dunes worldwide. Extracts from this plant have been previously revealed to have a variety of bioactive properties in humans. However, their effects on colorectal cancer cells remain poorly understood. In the present study, the potential biological activity of C. soldanella extracts in the colorectal cancer cell line HT-29 was examined. First, five solvent fractions [n-hexane, dichloromethane (DCM), ethyl acetate, n-butanol and water] were obtained from the crude extracts of C. soldanella through an organic solvent extraction method. In particular, the DCM fraction was demonstrated to exert marked dose- and time-dependent inhibitory effects according to results from the cell viability assay. Data obtained from the apoptosis assay suggested that the inhibition of HT-29 cell viability induced by DCM treatment was attributed to increased apoptosis. The apoptotic rate was markedly increased in a dose-dependent manner, which was associated with the protein expression levels of apoptosis-related proteins, including increased Fas, Bad and Bax, and decreased pro-caspase-8, Bcl-2, Bcl-xL, pro-caspase-9, pro-caspase-7 and pro-caspase-3. A mitochondrial membrane potential assay demonstrated that more cells became depolarized and the extent of cytochrome c release was markedly increased in a dose-dependent manner in HT-29 cells treated with DCM. In addition, cell cycle analysis confirmed S-phase arrest following DCM fraction treatment, which was associated with decreased protein expression levels of cell cycle-related proteins, such as cyclin A, CDK2, cell division cycle 25 A and cyclin dependent kinase inhibitor 1. Based on these results, the present study suggested that the DCM fraction of the C. soldanella extract can inhibit HT-29 cell viability whilst inducing apoptosis through mitochondrial membrane potential regulation and S-phase arrest. These results also suggested that the DCM fraction has potential anticancer activity in HT-29 colorectal cells. Further research on the composition of the DCM fraction is warranted.
Collapse
Affiliation(s)
- In-Hye Kim
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taekil Eom
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Joon-Young Park
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyung-Joo Kim
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taek-Jeong Nam
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| |
Collapse
|
8
|
Zhang MM, Li TJ, Liu JQ, Wang XS. Synthesis of 15-Arylisoquinolino[2′,1′:1,2] imidazo[4,5-f][1,10]phenanthrolines catalyzed by Copper(I)/o-Phen. RESEARCH ON CHEMICAL INTERMEDIATES 2021. [DOI: 10.1007/s11164-021-04395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
9
|
Potikha L, Brovarets V, Zhirnov V. Biological Evaluation of 3-Aminoisoquinolin-1(2H)-one Derivatives as Potential Anticancer Agents Authors Lyudmyla Potikha. FRENCH-UKRAINIAN JOURNAL OF CHEMISTRY 2021. [DOI: 10.17721/fujcv9i2p52-63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Anticancer activity of a series of 3-(hetaryl/aryl)amino substituted isoquinolin-1(2H)-ones has been studied within the international scientific program “NCI-60 Human Tumor Cell Lines Screen”. Screening was performed in vitro on 60 cell lines of lungs, kidneys, CNS, ovaries, prostate, and breast cancer, epithelial cancer, leukemia, and melanoma. The most effective compounds were those with thiazolyl or pyrazolyl substituent at 3-amino group and had no substituents at C(4) of the isoquinoline cycle. We identified a new lead compound, 3-(1,3-thiazol-2-ylamino)isoquinolin-1(2H)-one 12, which effectively prevents tumor cell growth (average lg GI50 = -5.18, lg TGI = -4.1, lg LC50 > -4.0) with good selectivity.
Collapse
|
10
|
Fouad MA, Abdel-Hamid H, Ayoup MS. Two decades of recent advances of Ugi reactions: synthetic and pharmaceutical applications. RSC Adv 2020; 10:42644-42681. [PMID: 35514898 PMCID: PMC9058431 DOI: 10.1039/d0ra07501a] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/24/2020] [Indexed: 12/30/2022] Open
Abstract
Multicomponent reactions (MCRs) are powerful synthetic tools in which more than two starting materials couple with each other to form multi-functionalized compounds in a one-pot process, the so-called "tandem", "domino" or "cascade" reaction, or utilizing an additional step without changing the solvent, the so-called a sequential-addition procedure, to limit the number of synthetic steps, while increasing the complexity and the molecular diversity, which are highly step-economical reactions. The Ugi reaction, one of the most common multicomponent reactions, has recently fascinated chemists with the high diversity brought by its four- or three-component-based isonitrile. The Ugi reaction has been introduced in organic synthesis as a novel, efficient and useful tool for the preparation of libraries of multifunctional peptides, natural products, and heterocyclic compounds with stereochemistry control. In this review, we highlight the recent advances of the Ugi reaction in the last two decades from 2000-2019, mainly in the synthesis of linear or cyclic peptides, heterocyclic compounds with versatile ring sizes, and natural products, as well as the enantioselective Ugi reactions. Meanwhile, the applications of these compounds in pharmaceutical trials are also discussed.
Collapse
Affiliation(s)
- Manar Ahmed Fouad
- Department of Chemistry, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Hamida Abdel-Hamid
- Department of Chemistry, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Mohammed Salah Ayoup
- Department of Chemistry, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| |
Collapse
|
11
|
A Novel Imidazopyridine Derivative Exerts Anticancer Activity by Inducing Mitochondrial Pathway-Mediated Apoptosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4929053. [PMID: 32908894 PMCID: PMC7468608 DOI: 10.1155/2020/4929053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/27/2020] [Indexed: 11/23/2022]
Abstract
Background Cancer remains a major clinical challenge because of the lack of effective drug for its treatment. To find out novel cancer chemotherapeutic molecules, we explored the anticancer effect of novel imidazopyridine compound 9i and also investigated the underlying molecular mechanism. Methods Human cervical cancer cell (HeLa) viability was measured by an MTT assay after treatment with compound 9i. Clonogenicity of HeLa cells was investigated by an in vitro colony formation assay. Cell death was visualized by propidium iodide (PI) staining. Fluorescence-activated cell sorting (FACS) was used to determine apoptosis and mitochondrial membrane potential in HeLa cells. The expression level of apoptosis-related proteins was also determined by western blot. Results Compound 9i suppressed HeLa cell viability in a time- and dose-dependent manner. Compound 9i induced mitochondrial outer membrane permeabilization (MOMP), activated caspase cascade, and finally resulted in apoptosis. Conclusion Compound 9i induces mitochondrial pathway-mediated apoptosis in human cervical cancer cells, suggesting that 9i could be a potential lead compound to be developed as a cancer therapeutic molecule.
Collapse
|
12
|
Chen J, Zhang L, Zheng X, Zhou J, Zhong T, Yu C. Synthesis of isoquinolinone derivatives by Rh (III)-catalyzed C–H functionalization of N-ethoxybenzamides. SYNTHETIC COMMUN 2020. [DOI: 10.1080/00397911.2020.1755984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Junyu Chen
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| | - Lei Zhang
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| | - Xiangyun Zheng
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| | - Jian Zhou
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| | - Tianshuo Zhong
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| | - Chuanming Yu
- College of pharmaceutical sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. of China
| |
Collapse
|
13
|
Tao Y, Hao X, Ding X, Cherukupalli S, Song Y, Liu X, Zhan P. Medicinal chemistry insights into novel CDC25 inhibitors. Eur J Med Chem 2020; 201:112374. [PMID: 32603979 DOI: 10.1016/j.ejmech.2020.112374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
Cell division cycle 25 (CDC25) phosphatases, a kind of cell cycle regulators, have become an attractive target for drug discovery, as they have been found to be over-expressed in various human cancer cells. Several CDC25 inhibitors have achieved significant attention in clinical trials with possible mechanistic actions. Prompted by the significance of CDC25 inhibitors with medicinal chemistry prospect, it is an apt time to review the various drug discovery methods involved in CDC25 drug discovery including high throughput screening (HTS), virtual screening (VS), fragment-based drug design, substitution decorating approach, structural simplification approach and scaffold hopping method to seek trends and identify promising new avenues of CDC25 drug discovery.
Collapse
Affiliation(s)
- Yucen Tao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Xia Hao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Xiao Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Srinivasulu Cherukupalli
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Yuning Song
- Department of Clinical Pharmacy, Qilu Hospital of Shandong University, 250012, Jinan, China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| |
Collapse
|
14
|
Kala P, Khasim Sharif S, Murali Krishna CH, Ramachandran D. Design, synthesis, and anticancer evaluation of 1,2,4-oxadiazole functionalized quinoline derivatives. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02467-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Cerchia C, Nasso R, Mori M, Villa S, Gelain A, Capasso A, Aliotta F, Simonetti M, Rullo R, Masullo M, De Vendittis E, Ruocco MR, Lavecchia A. Discovery of Novel Naphthylphenylketone and Naphthylphenylamine Derivatives as Cell Division Cycle 25B (CDC25B) Phosphatase Inhibitors: Design, Synthesis, Inhibition Mechanism, and in Vitro Efficacy against Melanoma Cell Lines. J Med Chem 2019; 62:7089-7110. [PMID: 31294975 DOI: 10.1021/acs.jmedchem.9b00632] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
CDC25 phosphatases play a critical role in the regulation of the cell cycle and thus represent attractive cancer therapeutic targets. We previously discovered the 4-(2-carboxybenzoyl)phthalic acid (NSC28620) as a new CDC25 inhibitor endowed with promising anticancer activity in breast, prostate, and leukemia cells. Herein, we report a structure-based optimization of NSC28620, leading to the identification of a series of novel naphthylphenylketone and naphthylphenylamine derivatives as CDC25B inhibitors. Compounds 7j, 7i, 6e, 7f, and 3 showed higher inhibitory activity than the initial lead, with Ki values in the low micromolar range. Kinetic analysis, intrinsic fluorescence studies, and induced fit docking simulations provided a mechanistic understanding of the activity of these derivatives. All compounds were tested in the highly aggressive human melanoma cell lines A2058 and A375. Compound 4a potently inhibited cell proliferation and colony formation, causing an increase of the G2/M phase and a reduction of the G0/G1 phase of the cell cycle in both cell lines.
Collapse
Affiliation(s)
- Carmen Cerchia
- Department of Pharmacy, "Drug Discovery" Laboratory , University of Naples Federico II , Via D. Montesano, 49 , 80131 Naples , Italy
| | - Rosarita Nasso
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy.,Department of Movement Sciences and Wellness , University of Naples "Parthenope" , 80133 Naples , Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences , University of Milan , Via Mangiagalli, 25 , 20133 Milan , Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences , University of Milan , Via Mangiagalli, 25 , 20133 Milan , Italy
| | - Arianna Gelain
- Department of Pharmaceutical Sciences , University of Milan , Via Mangiagalli, 25 , 20133 Milan , Italy
| | - Alessandra Capasso
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy
| | - Federica Aliotta
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy
| | - Martina Simonetti
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy
| | - Rosario Rullo
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy.,Institute for the Animal Production Systems in the Mediterranean Environment , Via Argine 1085 , 80147 Naples , Italy
| | - Mariorosario Masullo
- Department of Movement Sciences and Wellness , University of Naples "Parthenope" , 80133 Naples , Italy
| | - Emmanuele De Vendittis
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Via S. Pansini 5 , 80131 Naples , Italy
| | - Antonio Lavecchia
- Department of Pharmacy, "Drug Discovery" Laboratory , University of Naples Federico II , Via D. Montesano, 49 , 80131 Naples , Italy
| |
Collapse
|
16
|
Facile construction of fused benzimidazole-isoquinolinones that induce cell-cycle arrest and apoptosis in colorectal cancer cells. Bioorg Med Chem 2018; 26:3899-3908. [PMID: 29921474 DOI: 10.1016/j.bmc.2018.06.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/28/2018] [Accepted: 06/07/2018] [Indexed: 11/21/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequent, malignant gastrointestinal tumors, and strategies and effectiveness of current therapy are limited. A series of benzimidazole-isoquinolinone derivatives (BIDs) was synthesized and screened to identify novel scaffolds for CRC. Of the compounds evaluated, 7g exhibited the most promising anti-cancer properties. Employing two CRC cell lines, SW620 and HT29, 7g was found to suppress growth and proliferation of the cell lines at a concentration of ∼20 µM. Treatment followed an increase in G2/M cell cycle arrest, which was attributed to cyclin B1 and cyclin-dependent kinase 1 (CDK1) signaling deficiencies with simultaneous enhancement in p21 and p53 activity. In addition, mitochondrial-mediated apoptosis was induced in CRC cells. Interestingly, 7g decreased phosphorylated AKT, mTOR and 4E-BP1 levels, while promoting the expression/stability of PTEN. Since PTEN controls input into the PI3K/AKT/mTOR pathway, antiproliferative effects can be attributed to PTEN-mediated tumor suppression. Collectively, these results suggest that BIDs exert antitumor activity in CRC by impairing PI3K/AKT/mTOR signaling. Against a small kinase panel, 7g exhibited low affinity at 5 µM suggesting anticancer properties likely stem through a non-kinase mechanism. Because of the novelty of BIDs, the structure can serve as a lead scaffold to design new CRC therapies.
Collapse
|
17
|
Liu J, Wang YL, Zhang JH, Yang JS, Mou HC, Lin J, Yan SJ. Phosphatase CDC25B Inhibitors Produced by Basic Alumina-Supported One-Pot Gram-Scale Synthesis of Fluorinated 2-Alkylthio-4-aminoquinazolines Using Microwave Irradiation. ACS OMEGA 2018; 3:4534-4544. [PMID: 30221227 PMCID: PMC6130795 DOI: 10.1021/acsomega.8b00640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/17/2018] [Indexed: 06/08/2023]
Abstract
An efficient, environmentally benign, and inexpensive procedure has been developed for the synthesis of fluorinated 2-alkylthio-4-aminoquinazolines by microwave irradiation using basic alumina as a solid-support agent as well as a solid base. Notably, this protocol features improved energy efficiency, broad isothiourea substrate scope, easily available starting materials, and high atom efficiency and applicability toward gram-scale synthesis. Additionally, the target compounds were evaluated for the cytotoxic effect against human colon adenocarcinoma (HCT116 and HT29), human gastric cancer (SGC-7901), human lung adenocarcinoma (A549), and human hepatocyte carcinoma (HepG2) cells, and it was found that these compounds have excellent antitumor activities. Among them, compound 3e was found to be one of the most potent derivatives with IC50 values lower than 9.44 μM against five human tumor cell lines, making it more active than cisplatin (DDP). Furthermore, for the first time, the fluorinated 2-alkylthio-substituted 4-aminoquinazolines were identified as phosphatase CDC25B inhibitors.
Collapse
Affiliation(s)
- Jin Liu
- Key
Laboratory of Medicinal Chemistry for Natural Resource, Ministry of
Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R.
China
| | - Yu-Ling Wang
- Faculty
of Life Science and Technology, Kunming
University of Science and Technology, Kunming 650504, P. R. China
| | - Ji-Hong Zhang
- Faculty
of Life Science and Technology, Kunming
University of Science and Technology, Kunming 650504, P. R. China
| | - Jian-Shan Yang
- Key
Laboratory of Medicinal Chemistry for Natural Resource, Ministry of
Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R.
China
| | - Han-Chuan Mou
- Faculty
of Life Science and Technology, Kunming
University of Science and Technology, Kunming 650504, P. R. China
| | - Jun Lin
- Key
Laboratory of Medicinal Chemistry for Natural Resource, Ministry of
Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R.
China
| | - Sheng-Jiao Yan
- Key
Laboratory of Medicinal Chemistry for Natural Resource, Ministry of
Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R.
China
| |
Collapse
|
18
|
The design of novel inhibitors for treating cancer by targeting CDC25B through disruption of CDC25B-CDK2/Cyclin A interaction using computational approaches. Oncotarget 2018; 8:33225-33240. [PMID: 28402259 PMCID: PMC5464863 DOI: 10.18632/oncotarget.16600] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/17/2017] [Indexed: 01/28/2023] Open
Abstract
Cell division cycle 25B is a key cell cycle regulator and widely considered as potent clinical drug target for cancers. This research focused on identifying potential compounds in theory which are able to disrupt transient interactions between CDC25B and its CDK2/Cyclin A substrate.By using the method of ZDOCK and RDOCK, the most optimized 3D structure of CDK2/Cyclin A in complex with CDC25B was constructed and validated using two methods: 1) the superimposition of proteins; 2) analysis of the hydrogen bond distances of Arg 488(N1)-Asp 206(OD1), Arg 492(NE)-Asp 206(OD1), Arg 492(N1)-Asp 206(OD2) and Tyr 497(NE)-Asp 210(OD1). A series of new compounds was gained through searching the fragment database derived from ZINC based on the known inhibitor-compound 7 by the means of "replace fragment" technique. The compounds acquired via meeting the requirements of the absorption, distribution, metabolism, and excretion (ADME) predictions. Finally, 12 compounds with better binding affinity were identified. The comp#1, as a representative, was selected to be synthesized and assayed for their CDC25B inhibitory activities. The comp#1 exhibited mild inhibitory activities against human CDC25B with IC50 values at about 39.02 μM. Molecular Dynamic (MD) simulation revealed that the new inhibitor-comp#1 had favorable conformations for binding to CDC25B and disturbing the interactions between CDC25B and CDK2/Cyclin A.
Collapse
|
19
|
Salamoun JM, McQueeney KE, Patil K, Geib SJ, Sharlow ER, Lazo JS, Wipf P. Photooxygenation of an amino-thienopyridone yields a more potent PTP4A3 inhibitor. Org Biomol Chem 2018; 14:6398-402. [PMID: 27291491 DOI: 10.1039/c6ob00946h] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The phosphatase PTP4A3 is an attractive anticancer target, but knowledge of its exact role in cells remains incomplete. A potent, structurally novel inhibitor of the PTP4A family was obtained by photooxygenation of a less active, electron-rich thienopyridone (1). Iminothienopyridinedione 13 displays increased solution stability and is readily obtained by two new synthetic routes that converge in the preparation of 1. The late-stage photooxygenation of 1 to give 13 in high yield highlights the potential of this reaction to modify the structure and properties of a biological lead compound and generate value for expanding the scope of an SAR investigation. Analog 13 should become a valuable tool for further exploration of the role of PTP4A3 in tumor progression.
Collapse
Affiliation(s)
- Joseph M Salamoun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Kelley E McQueeney
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - Kalyani Patil
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Steven J Geib
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Elizabeth R Sharlow
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - John S Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| |
Collapse
|
20
|
McQueeney KE, Salamoun JM, Burnett JC, Barabutis N, Pekic P, Lewandowski SL, Llaneza DC, Cornelison R, Bai Y, Zhang ZY, Catravas JD, Landen CN, Wipf P, Lazo JS, Sharlow ER. Targeting ovarian cancer and endothelium with an allosteric PTP4A3 phosphatase inhibitor. Oncotarget 2018; 9:8223-8240. [PMID: 29492190 PMCID: PMC5823565 DOI: 10.18632/oncotarget.23787] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/25/2017] [Indexed: 12/16/2022] Open
Abstract
Overexpression of protein tyrosine phosphatase PTP4A oncoproteins is common in many human cancers and is associated with poor patient prognosis and survival. We observed elevated levels of PTP4A3 phosphatase in 79% of human ovarian tumor samples, with significant overexpression in tumor endothelium and pericytes. Furthermore, PTP4A phosphatases appear to regulate several key malignant processes, such as invasion, migration, and angiogenesis, suggesting a pivotal regulatory role in cancer and endothelial signaling pathways. While phosphatases are attractive therapeutic targets, they have been poorly investigated because of a lack of potent and selective chemical probes. In this study, we disclose that a potent, selective, reversible, and noncompetitive PTP4A inhibitor, JMS-053, markedly enhanced microvascular barrier function after exposure of endothelial cells to vascular endothelial growth factor or lipopolysaccharide. JMS-053 also blocked the concomitant increase in RhoA activation and loss of Rac1. In human ovarian cancer cells, JMS-053 impeded migration, disrupted spheroid growth, and decreased RhoA activity. Importantly, JMS-053 displayed anticancer activity in a murine xenograft model of drug resistant human ovarian cancer. These data demonstrate that PTP4A phosphatases can be targeted in both endothelial and ovarian cancer cells, and confirm that RhoA signaling cascades are regulated by the PTP4A family.
Collapse
Affiliation(s)
- Kelley E. McQueeney
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | | | - James C. Burnett
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nektarios Barabutis
- Frank Reidy Center for Bioelectrics, Old Dominion University, Norfolk, VA, USA
| | - Paula Pekic
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | | | - Danielle C. Llaneza
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Robert Cornelison
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Yunpeng Bai
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - John D. Catravas
- Frank Reidy Center for Bioelectrics, Old Dominion University, Norfolk, VA, USA
| | - Charles N. Landen
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - John S. Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
21
|
Zhang L, Zheng X, Chen J, Cheng K, Jin L, Jiang X, Yu C. Ru(ii)-Catalyzed C6-selective C–H amidation of 2-pyridones. Org Chem Front 2018. [DOI: 10.1039/c8qo00795k] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An efficient Ru-catalyzed C6 site-selective amidation of 2-pyridones has been accomplished with dioxazolone under mild conditions.
Collapse
Affiliation(s)
- Lei Zhang
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Xiangyun Zheng
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Jinkang Chen
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Kang Cheng
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Licheng Jin
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Xinpeng Jiang
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Chuanming Yu
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals
| |
Collapse
|
22
|
Liu S, Cai X, Xia L, Jiang C, Chen P, Wang X, Zhang B, Zhao HY. Chloroquine exerts antitumor effects on NB4 acute promyelocytic leukemia cells and functions synergistically with arsenic trioxide. Oncol Lett 2017; 15:2024-2030. [PMID: 29434902 DOI: 10.3892/ol.2017.7488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 10/26/2017] [Indexed: 01/17/2023] Open
Abstract
Chloroquine (CQ) has been confirmed to exhibit antitumor effects on different types of cancer cell, but whether it exerts the same effect on acute promyelocytic leukemia (APL) cells remains to be confirmed. In the present study, the effects of various concentrations of CQ on the growth, apoptosis and cell cycle distribution of NB4 cells, as well as the potential mechanisms underlying these effects, were examined. The combined effect of CQ and arsenic trioxide (ATO) on the growth of NB4 cells was also determined. The results of the present study demonstrated that CQ treatment inhibited cell proliferation, and induced mitochondrial pathway apoptosis and S phase arrest in a dose-dependent manner by regulating apoptosis- and cell cycle-related proteins. CQ and ATO had a synergistic effect on the growth inhibition of NB4 cells, which may have been induced through the inhibition of autophagy. In conclusion, the results of the present study indicated that CQ exhibits a cytotoxic effect on NB4 cells and has a synergistic effect when combined with ATO, which thereby improves the curative effect of ATO on APL.
Collapse
Affiliation(s)
- Shousheng Liu
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiuyu Cai
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Liangping Xia
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Chang Jiang
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Ping Chen
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaopai Wang
- Department of Pathology, Guangzhou First People's Hospital, Guangzhou, Guangdong 510080, P.R. China
| | - Bei Zhang
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Hong Yun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
23
|
Small molecule targeting of PTPs in cancer. Int J Biochem Cell Biol 2017; 96:171-181. [PMID: 28943273 DOI: 10.1016/j.biocel.2017.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 01/28/2023]
Abstract
Protein tyrosine phosphatases (PTPs) undeniably have a central role in the development and progression of human cancers. Historically, however, PTPs have not been viewed as privileged drug targets, and progress on identifying potent, selective, and cell-active small molecule PTP inhibitors has suffered accordingly. This situation is rapidly changing, however, due to biochemical advances in the study of PTPs and recent small molecule screening campaigns, which have identified potent and mechanistically diverse lead structures. These compounds are facilitating the exploration of the fundamental cellular processes controlled by PTPs in cancers, and could form the inflection point for new therapeutic paradigms for the treatment of a range of cancers. Herein, we review recent advances in the discovery and biological annotation of cancer-relevant small molecule PTP inhibitors.
Collapse
|
24
|
Lazo JS, McQueeney KE, Sharlow ER. New Approaches to Difficult Drug Targets: The Phosphatase Story. SLAS DISCOVERY 2017; 22:1071-1083. [DOI: 10.1177/2472555217721142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The drug discovery landscape is littered with promising therapeutic targets that have been abandoned because of insufficient validation, historical screening failures, and inferior chemotypes. Molecular targets once labeled as “undruggable” or “intractable” are now being more carefully interrogated, and while they remain challenging, many target classes are appearing more approachable. Protein tyrosine phosphatases represent an excellent example of a category of molecular targets that have emerged as druggable, with several small molecules and antibodies recently becoming available for further development. In this review, we examine some of the diseases that are associated with protein tyrosine phosphatase dysfunction and use some prototype contemporary strategies to illustrate approaches that are being used to identify small molecules targeting this enzyme class.
Collapse
Affiliation(s)
- John S. Lazo
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| | - Kelley E. McQueeney
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth R. Sharlow
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
25
|
Pterostilbene Induces Cell Apoptosis and Cell Cycle Arrest in T-Cell Leukemia/Lymphoma by Suppressing the ERK1/2 Pathway. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9872073. [PMID: 28785594 PMCID: PMC5529638 DOI: 10.1155/2017/9872073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/28/2017] [Accepted: 05/29/2017] [Indexed: 12/22/2022]
Abstract
Pterostilbene is a natural 3,5-dimethoxy analog of trans-resveratrol that has been reported to have antitumor, antioxidant, and anti-inflammatory effects. T-cell leukemia/lymphoma is one of the more aggressive yet uncommon non-Hodgkin lymphomas. Although there has been increasing research into T-cell leukemia/lymphoma, the molecular mechanisms of the antitumor effects of pterostilbene against this malignancy are still largely unknown. The aim of this study is to confirm the effects of pterostilbene in T-cell leukemia/lymphoma. Jurkat and Hut-78 cells treated with pterostilbene were evaluated for cell proliferation using Cell Counting Kit-8, and apoptosis, cell cycle progression, reactive oxygen species generation, and mitochondrial membrane potential were analyzed using flow cytometry. The level of protein expression was detected by western blot. The results demonstrated that pterostilbene significantly inhibited the growth of T-cell leukemia/lymphoma cell lines in vitro and induced apoptosis in a dose- and time-dependent manner. Moreover, pterostilbene treatment markedly induced S-phase cell cycle arrest, which was accompanied by downregulation of cdc25A, cyclin A2, and CDK2. Pterostilbene also induced the generation of reactive oxygen species and the loss of mitochondrial membrane potential and inhibited ERK1/2 phosphorylation. Taken together, our study demonstrated the potential of pterostilbene to be an effective treatment for T-cell leukemia/lymphoma.
Collapse
|
26
|
Lindsay-Scott PJ, Gallagher PT. Synthesis of heterocycles from arylacetonitriles: Powerful tools for medicinal chemists. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.05.089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Zwergel C, Czepukojc B, Evain-Bana E, Xu Z, Stazi G, Mori M, Patsilinakos A, Mai A, Botta B, Ragno R, Bagrel D, Kirsch G, Meiser P, Jacob C, Montenarh M, Valente S. Novel coumarin- and quinolinone-based polycycles as cell division cycle 25-A and -C phosphatases inhibitors induce proliferation arrest and apoptosis in cancer cells. Eur J Med Chem 2017; 134:316-333. [DOI: 10.1016/j.ejmech.2017.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/06/2023]
|
28
|
Lee JI, Kim IH, Nam TJ. Crude extract and solvent fractions of Calystegia soldanella induce G1 and S phase arrest of the cell cycle in HepG2 cells. Int J Oncol 2017; 50:414-420. [PMID: 28101580 PMCID: PMC5238786 DOI: 10.3892/ijo.2017.3836] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 12/23/2016] [Indexed: 11/06/2022] Open
Abstract
The representative halophyte Calystegia soldanella (L) Roem. et Schult is a perennial vine herb that grows in coastal dunes throughout South Korea as well as in other regions around the world. This plant has long been used as an edible and medicinal herb to cure rheumatic arthritis, sore throat, dropsy, and scurvy. Some studies have also shown that this plant species exhibits various biological activities. However, there are few studies on cytotoxicity induced by C. soldanella treatment in HepG2 human hepatocellular carcinoma cells. In this study, we investigated the viability of HepG2 cells following treatment with crude extracts and four solvent-partitioned fractions of C. soldanella. Of the crude extract and four solvent fractions tested, treatment with the 85% aqueous methanol (aq. MeOH) fraction resulted in the greatest inhibition of HepG2 cell proliferation. Flow cytometry showed that the 85% aq. MeOH fraction induced a G0/G1 and S phase arrest of the cell cycle progression. The 85% aq. MeOH fraction arrested HepG2 cells at the G0/G1 phase in a concentration-dependent manner, and resulted in decreased expression of cyclin D1, cyclin E, cyclin-dependent kinase (CDK)2, CDK4, CDK6, p21, and p27. Additionally, the 85% aq. MeOH fraction treatment also arrested HepG2 cells in the S phase, with decreased expression of cyclin A, CDK2, and CDC25A. Also, treatment with this fraction reduced the expression of retinoblastoma (RB) protein and the transcription factor E2F. These results suggest that the 85% aq. MeOH fraction exhibits potential anticancer activity in HepG2 cells by inducing G0/G1 and S phase arrest of the cell cycle.
Collapse
Affiliation(s)
- Jung Im Lee
- Institute of Fisheries Science, Pukyong National University, Ilgwang-myeon, Gijang-gun, Busan 619-911
| | - In-Hye Kim
- Institute of Fisheries Science, Pukyong National University, Ilgwang-myeon, Gijang-gun, Busan 619-911
| | - Taek-Jeong Nam
- Institute of Fisheries Science, Pukyong National University, Ilgwang-myeon, Gijang-gun, Busan 619-911
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea
| |
Collapse
|
29
|
R-Phycoerythrin Induces SGC-7901 Apoptosis by Arresting Cell Cycle at S Phase. Mar Drugs 2016; 14:md14090166. [PMID: 27626431 PMCID: PMC5039537 DOI: 10.3390/md14090166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/03/2016] [Accepted: 09/07/2016] [Indexed: 02/04/2023] Open
Abstract
R-Phycoerythrin (R-PE), one of the chemical constituents of red algae, could produce singlet oxygen upon excitation with the appropriate radiation and possibly be used in photodynamic therapy (PDT) for cancer. Documents reported that R-PE could inhibit cell proliferation in HepG2 and A549 cells, which was significative for cancer therapy. This is due to the fact that R-PE could kill cancer cells directly as well as by PDT. However, little is known about the cytotoxicity of R-PE to the SGC-7901 cell. In this study, it has been found that R-PE could inhibit SGC-7901 proliferation and induce cell apoptosis, which was achieved by arresting the SGC-7901 cell at S phase. CyclinA, CDK2 and CDC25A are proteins associated with the S phase, and it was found that R-PE could increase the expression of cyclin A protein and decrease the expression of CDK2 and CDC25A proteins. Thus, it was concluded that R-PE reduced the CDK2 protein activated through decreasing the CDC25A factor, which reduced the formation of Cyclin-CDK complex. The reduction of Cyclin-CDK complex made the SGC-7901 cells arrest at the S phase. Therefore, R-PE induced apoptosis by arresting the SGC-7901 cell at S phase was successful, which was achieved by the expression of the CDC25A protein, which reduced the CDK2 protein actived and the formation of Cyclin-CDK complex.
Collapse
|
30
|
Abstract
Cancer, more than any other human disease, now has a surfeit of potential molecular targets poised for therapeutic exploitation. Currently, a number of attractive and validated cancer targets remain outside of the reach of pharmacological regulation. Some have been described as undruggable, at least by traditional strategies. In this article, we outline the basis for the undruggable moniker, propose a reclassification of these targets as undrugged, and highlight three general classes of this imposing group as exemplars with some attendant strategies currently being explored to reclassify them. Expanding the spectrum of disease-relevant targets to pharmacological manipulation is central to reducing cancer morbidity and mortality.
Collapse
Affiliation(s)
- John S Lazo
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908-0735; ,
| | - Elizabeth R Sharlow
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908-0735; ,
| |
Collapse
|
31
|
Jiang Y, Sun R, Wang Q, Tang XY, Shi M. Cyclization of sulfide, ether or tertiary amine-tethered N-sulfonyl-1,2,3-triazoles: a facile synthetic protocol for 3-substituted isoquinolines or dihydroisoquinolines. Chem Commun (Camb) 2015; 51:16968-71. [DOI: 10.1039/c5cc07511d] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A novel cyclization reaction of sulfide, ether or tertiary amine-tethered N-sulfonyl-1,2,3-triazoles, has been developed, efficiently affording 3-substituted isoquinoline or dihydroisoquinoline derivatives.
Collapse
Affiliation(s)
- Yu Jiang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals
- East China University of Science and Technology
- Shanghai
- China
| | - Run Sun
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals
- East China University of Science and Technology
- Shanghai
- China
| | - Qiang Wang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals
- East China University of Science and Technology
- Shanghai
- China
| | - Xiang-Ying Tang
- State Key Laboratory of Organometallic Chemistry
- Shanghai Institute of Organic Chemistry
- Chinese Academy of Sciences
- Shanghai 200032
- China
| | - Min Shi
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals
- East China University of Science and Technology
- Shanghai
- China
- State Key Laboratory of Organometallic Chemistry
| |
Collapse
|